WO2003048735A2 - Processes for the measurement of the potency of glatiramer acetate - Google Patents

Processes for the measurement of the potency of glatiramer acetate Download PDF

Info

Publication number
WO2003048735A2
WO2003048735A2 PCT/US2002/038859 US0238859W WO03048735A2 WO 2003048735 A2 WO2003048735 A2 WO 2003048735A2 US 0238859 W US0238859 W US 0238859W WO 03048735 A2 WO03048735 A2 WO 03048735A2
Authority
WO
WIPO (PCT)
Prior art keywords
batch
cells
glatiramer acetate
test
potency
Prior art date
Application number
PCT/US2002/038859
Other languages
English (en)
French (fr)
Other versions
WO2003048735A3 (en
Inventor
Ety Klinger
Original Assignee
Teva Pharmaceutical Industries, Ltd.
Teva Pharmaceuticals Usa, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=23326090&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2003048735(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to JP2003549881A priority Critical patent/JP4369234B2/ja
Priority to DK02790028.1T priority patent/DK1459065T3/da
Priority to KR1020047008518A priority patent/KR100657048B1/ko
Priority to IL16212702A priority patent/IL162127A0/xx
Priority to EP02790028A priority patent/EP1459065B1/en
Priority to DE60237170T priority patent/DE60237170D1/de
Priority to NZ533327A priority patent/NZ533327A/en
Priority to AU2002353059A priority patent/AU2002353059B2/en
Priority to CA2469393A priority patent/CA2469393C/en
Application filed by Teva Pharmaceutical Industries, Ltd., Teva Pharmaceuticals Usa, Inc. filed Critical Teva Pharmaceutical Industries, Ltd.
Priority to MXPA04005433A priority patent/MXPA04005433A/es
Priority to SI200230921T priority patent/SI1459065T1/sl
Priority to CNB028277732A priority patent/CN1308683C/zh
Priority to AT02790028T priority patent/ATE475883T1/de
Publication of WO2003048735A2 publication Critical patent/WO2003048735A2/en
Publication of WO2003048735A3 publication Critical patent/WO2003048735A3/en
Priority to IL162127A priority patent/IL162127A/en
Priority to IS7286A priority patent/IS7286A/is
Priority to ZA2004/04472A priority patent/ZA200404472B/en
Priority to NO20042816A priority patent/NO338247B1/no
Priority to HK05102405.0A priority patent/HK1070421A1/xx
Priority to IL198285A priority patent/IL198285A/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5038Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects involving detection of metabolites per se
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/02Peptides of undefined number of amino acids; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • G01N33/6866Interferon
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • G01N33/6869Interleukin
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/54Interleukins [IL]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/54Interleukins [IL]
    • G01N2333/5412IL-6
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/54Interleukins [IL]
    • G01N2333/5428IL-10
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/54Interleukins [IL]
    • G01N2333/55IL-2
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/555Interferons [IFN]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/555Interferons [IFN]
    • G01N2333/57IFN-gamma

Definitions

  • the present invention relates to methods of standardizing the measurement of the potency of glatiramer acetate based on specific recognition of the glatiramer acetate by T cells.
  • Glatiramer acetate (GA, also known as Copolymer-1 (Physician's Desk Reference), Copolymer 1, Cop-1 or COPAXONE ® ) , is an approved drug for the treatment of multiple sclerosis (MS) .
  • Glatiramer acetate consists of the acetate salts of synthetic polypeptides, containing four naturally occurring amino acids (Physician's Desk Reference): L-glutamic acid, L-alanine, L- tyrosine, and L-lysine (Physician's Desk Reference) with an average molar fraction of L-glutamic acid: 0.129-0.153; L- alanine: 0.392-0.462; L-tyrosine: 0.086-0.100; L-lysine: 0.300- 0.374, respectively.
  • glatiramer acetate The average molecular weight of glatiramer acetate is 4,700 - 11,000 daltons (Physician's Desk Reference). Chemically, glatiramer acetate is designated L-glutamic acid polymer with L-alanine, L-lysine and L-tyrosine, acetate (salt) (Physician's Desk Reference). Its structural formula is:
  • Glatiramer acetate is also written as: poly[L-Glu 13"15 , L-Ala 39"46 , L-Tyr 8 6"10 , L-Lys 30"37 ] -nCH 3 COOH.
  • Glatiramer acetate was shown to suppress experimental autoimmune encephalomyelitis (EAE) -- an experimental model for multiple sclerosis (MS) in various animal species (Lando et al . , 1979;
  • T cell activation It is commonly accepted that a high level of antigen specificity is a feature of T cell activation.
  • the T cells of the immune system recognize immunogenic peptides complexed to the major histocompatibility complex (MHC) class II or I molecules, expressed on antigen presenting cells (APCs) .
  • MHC major histocompatibility complex
  • APCs antigen presenting cells
  • the specificity of antigen recognition by T cells is defined by several parameters: 1) affinity of the T cell receptor to the MHC peptide complex; 2) primary sequence of the antigenic peptide; and 3) synergistic effects of certain amino acid combinations within the antigenic peptide.
  • affinity of the T cell receptor to the MHC peptide complex 2) primary sequence of the antigenic peptide
  • synergistic effects of certain amino acid combinations within the antigenic peptide Based on current knowledge on the mechanism of action of glatiramer acetate, it is believed that the biological activity of glatiramer acetate in MS is mediated by immuno
  • the subject invention provides a process for measuring the potency of a test batch of glatiramer acetate relative to the known potency of a reference batch of glatiramer acetate which comprises
  • step (b) separately incubating at least five reference samples, each of which contains a predetermined number of cells from the primary culture of step (b) and a predetermined amount of glatiramer acetate between 1 ⁇ g/ml and 25 ⁇ g/ml from a reference batch;
  • step (b) incubating at least two samples, each of which contains a predetermined number of cells from the primary culture of step (b) and a predetermined amount of glatiramer acetate from the test batch;
  • the predetermined number of cells is substantially identical, and wherein for each sample containing a predetermined amount of glatiramer acetate from the test batch there is a corresponding reference sample containing a substantially identical predetermined amount of glatiramer acetate from the reference batch.
  • the subject invention also provides a process for measuring the potency of a test batch of glatiramer acetate relative to the known potency of a reference batch of glatiramer acetate which comprises
  • step (b) preparing a primary culture of cells from the test mammal of step (a) at a predetermined time after immunization
  • step (b) separately incubating at least two reference samples, each of which contains a predetermined number of cells from the primary culture of step (b) and a predetermined amount of glatiramer acetate from a reference batch;
  • step (b) incubating at least two samples, each of which contains a predetermined number of cells from the primary culture of step (b) and a predetermined amount of glatiramer acetate from the test batch;
  • the predetermined number of cells is substantially identical, and wherein for each immunization sample containing a predetermined amount of glatiramer acetate from the test batch there is a corresponding reference sample containing a substantially identical predetermined amount of glatiramer acetate from the reference batch.
  • Figure 1 Immunization with GA RS (Reference Standard) . Primary culture of LN cells. IL-2 Detection by ELISA.
  • FIG. 2 Induction of GA-specific T cells.
  • Figure 3 Diagram of hemacytometer .
  • Figure 4 Optimization of the immunization protocol - culture source and GA Reference Standard (RS) dose effect.
  • Primary cultures of Lymph Node (LN) and spleen cells were derived from mice immunized with 10 or 250 ⁇ g GA RS + complete Freund's adjuvant (CFA) .
  • the cells were cultured in the presence of increasing concentrations of GA RS .
  • the culture media were collected and assayed for IL-2 by enzyme-linked immunoabsorbent assay (ELISA) .
  • ELISA enzyme-linked immunoabsorbent assay
  • Figure 5 Optimization of the immunization protocol - adjuvant and dose effect.
  • Primary cultures of LN cells derived from mice immunized with 250 ⁇ g GA RS + CFA or with 10 mg GA RS + incomplete Freund's adjuvant (ICFA) were cultured in the presence of increasing concentrations of GA RS .
  • ICFA incomplete Freund's adjuvant
  • Figure 6 Effect of the immunization period. Mice were immunized with 250 ⁇ g GA RS in CFA and LN were removed after 9, 10 and 11 days. The response of the LN cells from different groups to various concentrations of GA RS was tested in vi tro by measuring IL-2 secretion by ELISA.
  • Figure 7 Effect of the culture media on GA-specific T cell response.
  • Primary cultures of LN cells were cultured with different media containing either 1% normal mouse serum (NMS) , 1% fetal bovine serum (FBS) or defined cell culture media (DCCM1)
  • NMS normal mouse serum
  • FBS 1% fetal bovine serum
  • DCCM1 defined cell culture media
  • Figure 8 Kinetics of IL-2 secretion in response to GA RS .
  • a primary culture of LN cells was prepared from mice immunized with 250 ⁇ g GA RS + CFA. The cells were incubated with 0, 0.5, 2.5 and 5 ⁇ g/ml GA RS at 37°C for the indicated intervals. At each time point, an aliquot of 5 X 10 6 cells was centrifuged and the supernatant was kept at -20°C. All samples were assayed simultaneously for IL-2 by ELISA.
  • Figure 9 Stability of IL-2 in culture media.
  • a primary culture of LN cells was prepared from mice immunized with 250 ⁇ g GA RS + CFA. The cells were incubated with various concentrations of GA RS at 37°. After overnight incubation, the supernatants were collected and divided into two aliquots. One aliquot was assayed immediately by ELISA, and the second was kept for 7 days at -20°C prior to being assayed.
  • FIG. 10 Stability of GA RS solution at -20°C.
  • GA RS solution of 1 mg/ml was prepared, divided into aliquots and kept at -20°C.
  • the dose-response of the GA-specific cells to GA RS solution was tested at time zero (Date 1) and after 5 months at -20°C.
  • Figure 11 Effect of the average molecular weight (MW) on the GA RS-specific T-cell response.
  • a primary culture of LN cells was prepared from mice immunized with 250 ⁇ g GA RS + CFA. The cells were cultured in the presence of 2 different concentrations of GA RS and of GA Drug Substance (DS) of different molecular weights. Following overnight incubation at 37°C the culture media were collected and assayed for IL-2 by ELISA.
  • MW average molecular weight
  • Figure 12 (A & B) : Cross-reactivity of GA RS-specific T cells with GA DS and Drug Product (DP) batches.
  • a primary culture of LN cells was prepared from mice immunized with 250 ⁇ g GA RS + CFA.
  • the response of the GA RS-specific T cells to another GA DS batch (Fig. 12A) , to a GA DP batch and to mannitol (Fig. 12B) was compared to the response to the GA RS batch.
  • IL-2 levels in the culture media were measured by ELISA.
  • FIG. 13 Kinetics of GA RS proteolysis by trypsin.
  • GA RS was proteolysed by trypsin for the indicated time points. The activity of the proteolysed samples was tested by the in vi tro potency test .
  • FIG. 14 Reverse-Phase High Pressure Liquid Chromatography (RP- HPLC) of GA before and after proteolysis by trypsin.
  • GA RS was proteolysed by trypsin and the chromatographic profile of the samples was tested by RP-HPLC.
  • FIG. 15 Kinetics of GA RS proteolysis by chymotrypsin.
  • GA RS was proteolysed by chymotrypsin for the indicated time points. The activity of the proteolysed samples was tested by the in vi tro potency test.
  • FIG. 16 RP-HPLC of GA before and after proteolysis by chymotrypsin.
  • GA RS was proteolysed by chymotrypsin and the chromatographic profile of the samples was tested by RP-HPLC.
  • the subject invention provides a process for measuring the potency of a test batch of glatiramer acetate relative to the known potency of a reference batch of glatiramer acetate which comprises
  • step (b) separately incubating at least five reference samples, each of which contains a predetermined number of cells from the primary culture of step (b) and a predetermined amount of glatiramer acetate between 1 ⁇ g/ml' and 25 ⁇ g/ml from a reference batch;
  • step (b) incubating at least two samples, each of which contains a predetermined number of cells from the primary culture of step (b) and a predetermined amount of glatiramer acetate from the test batch;
  • the predetermined number of cells is substantially identical, and wherein for each sample containing a predetermined amount of glatiramer acetate from the test batch there is a corresponding reference sample containing a substantially identical predetermined amount of glatiramer acetate from the reference batch.
  • six reference samples are separately incubated in step (d) .
  • the subject invention also provides a process for measuring the potency of a test batch of glatiramer acetate relative to the known potency of a reference batch of glatiramer acetate which comprises
  • step (b) preparing a primary culture of cells from the test mammal of step (a) at a predetermined time after immunization
  • step (b) separately incubating at least two reference samples, each of which contains a predetermined number of cells from the primary culture of step (b) and a predetermined amount of glatiramer acetate from a reference batch;
  • step (b) incubating at least two samples, each of which contains a predetermined number of cells from the primary culture of step (b) and a predetermined amount of glatiramer acetate from the test batch; e. determining for each sample in steps (c) and (d) , the amount of a cytokine secreted by the cells in each sample after a predetermined time period of incubation of such sample;
  • the predetermined number of cells is substantially identical, and wherein for each immunization sample containing a predetermined amount of glatiramer acetate from the test batch there is a corresponding reference sample containing a substantially identical predetermined amount of glatiramer acetate from the reference batch.
  • the cytokine is an interleukin.
  • the interleukin is interleukin-2.
  • the interleukin is interleukin- 6.
  • the interleukin is interleukin-10.
  • the cytokine is interferon-gamma .
  • the mammal produces T cells specific to glatiramer acetate reference standard. In another embodiment, the mammal is a rodent.
  • the rodent is a mouse.
  • the mouse is a female (SJLXBALB/C) Fl mouse .
  • the mammal is about 8 to about 12 weeks old.
  • the cells are lymph node cells.
  • the cells are spleen cells.
  • the subject invention further provides a process for preparing a batch of glatiramer acetate as acceptable for pharmaceutical use which comprises
  • the subject invention provides a process for preparing glatiramer acetate acceptable for pharmaceutical use which comprises
  • the present invention provides the standardization of the measurement of the potency of GA.
  • the potency test quantitatively determines the biological activity of GA. This is the first showing ever of such a test.
  • This standardization method is essential in order to show batch to batch reproducibility with regards to potency and quality of DS and DP.
  • DS refers to the active ingredient, i.e., GA.
  • DP is used to indicate the finished product, i.e., Copaxone ® .
  • RS denotes a batch of glatiramer acetate having an average molecular weight of about 7000 Da.
  • the subject invention makes use of the observation that T cells incubated with a cytokine, e.g., IL-2, proliferate in response to that cytokine (Lisak et al . , 1974) .
  • a cytokine e.g., IL-2
  • GA RS was produced as described in U.S. Patent No. 5,800,808 or PCT International Publication No. WO 00/05250.
  • the GA RS was chosen based on the chemical and biological properties being in the midrange of Copaxone ® as described above.
  • a primary culture of LN cells was prepared, and the cells were incubated with various concentrations of GA RS and with test samples.
  • the culture media were collected and the level of IL-2 was measured by ELISA.
  • the T-cell response to each DS batch were tested at two concentrations (within the linear range) , and the % potency of the DS batch was calculated relative to that of the GA RS batch.
  • the purpose of this procedure was to determine the relative potency of GA DS batch in vi tro, using GA RS-specific T cells.
  • IL-2 was measured by ELISA kit: OptEIATM Set: mouse IL-2 (Pharmingen, Cat. # 2614KI, or equivalent).
  • Con A solution Primary bank The contents of one vial of 5 mg Con were dissolved in 1 ml PBS, mixed well and divided into aliquots of 50 ⁇ l each. The aliquots were kept at -20°C up to the expiration date set by the manufacturer .
  • Secondary bank One aliquot of the primary bank (5 mg/ml) was thawed and diluted with 4.95 ml of enriched DCCM1 medium to obtain a solution of 50 ⁇ g/ml . The solution was divided into working aliquots of 100 ⁇ l each and kept at -20°C. Upon use, one aliquot from the secondary bank was thawed and diluted up to 1 ml (a 10-fold dilution) with enriched DCCM1 to obtain a solution of 5 ⁇ g/ml Con A.
  • GA RS emulsion in CFA was prepared under sterile conditions, i.e., in a laminar hood, using sterile equipment and materials.
  • the GA RS emulsion was transferred into an insulin syringe. Then, 100 ⁇ l of the emulsion (250 ⁇ g GA per mouse) were injected into four footpads of each naive mouse (about 25 ⁇ l into each footpad) . The immunized mice were used for the in vi tro test 9-11 days following immunization.
  • the primary culture of LN cells was prepared 9-11 days following immunization, according to the following procedure:
  • the UN lamp was turned on 20 minutes before commencing work in the laminar hood and turned off when work began. Prior to placing any reagents under the hood, the working surface was cleaned with a 70% ethanol solution. Enriched DCCM1 medium was prepared. The enriched DCCM1 and the RPMI medium were pre-warmed at 37°C prior to use. The mice were sacrificed by cervical dislocation. Each mouse was placed on its back and fastened to a support platform. The abdomen was sprayed with 70% alcohol and a middle incision was made (a 2 cm-long incision was usually sufficient) . The skin was intersected towards the hind legs and L ⁇ was located from the hind and forelegs .
  • the L ⁇ was transferred into a sterile petri dish containing about 5 ml sterile RPMI medium and the L ⁇ cells were teased out by a sterile syringe plunger.
  • the sterile syringe was used to collect the cells' suspension from the petri dish (the collection of tissue debris was avoided by using sterile needles) .
  • the cells' suspension were transferred into a 50 ml sterile tube.
  • Cell counting Example procedure for counting LN cells derived from 5 immunized mice
  • the cells' tube were filled with RPMI medium up to 40 ml.
  • the LN cells were centrifuged at 200 xg for 10 minutes at room temperature (15-25°C) .
  • the pellet was re-suspended with 40 ml RPMI.
  • Two aliquots of 50 ⁇ l were drawn each from the cells' suspension diluted 4 -fold with 150 ⁇ l of 0.1% Trypan blue in a microtest well. The aliquots were mixed well by pipetting gently up and down.
  • the hemacytometer was covered with a cover slip.
  • a 50-200 ⁇ l pipettman was used to load both aliquots into the upper and lower chambers of the hemacytometer, one suspension in each chamber. The mixture was allowed to settle within the chambers for about 2 minutes.
  • the viable cells were counted in the central square (composed of 25 large squares of 16 small squares each, see Fig. 3) of the upper and lower chambers.
  • the viable cells did not absorb Trypan blue and were therefore characterized by a clear appearance. However, dead cells were permeable to Trypan blue and appeared blue in color.
  • Cells appearing on the border of the central square were only counted if a portion of the cell was actually within the central square. If a cell was on the border, and not at all within the central square, it was not counted.
  • the cells' density was calculated using the following equation:
  • Average number viable cells (both chambers) x 4 x 10 4 cells ml
  • the cells were centrifuged at 200 xg for 10 minutes at room temperature.
  • the cells were re-suspended to a density of lxlO 7 cells/ml with enriched DCCMl.
  • the in vitro bioassay was performed in a 24 -well, flat -bottomed tissue culture test plate at a final volume of 1 ml.
  • the GA RS stock solution was diluted to 100 ⁇ g/ml (10-fold) with enriched DCCMl medium and filtered through a 0.2 ⁇ cellulose acetate filter.
  • Six serial dilutions of the GA RS solution with enriched DCCMl medium were prepared between 2-50 ⁇ g/ml, as described by the example in Table 2.
  • LN cells final density, for example, 5xl0 6 cells/well
  • the density of the cells was changed depending upon their response to GA.
  • the cultures were kept at 37°C in a humidified 5% C0 2 incubator for 18-21 hrs.
  • the plate was centrifuged at 200 xg for 10 minutes at room temperature.
  • the supernatants were collected into cryotubes.
  • the supernatants were divided into working aliquots to avoid repeated freezing/thawing of the samples.
  • the supernatants were stored at -20°C for up to one week.
  • the hood was cleaned with 70% ethanol solution and dried with Kim wipes. The gloves were removed and the hands were immediately washed with disinfectant.
  • the culture media of GA RS, test samples and controls were diluted with enriched DCCMl as follows: a) A 2-fold dilution of the 1 and 2.5 ⁇ g/ml GA RS sample and of the negative control sample; b) 5-10 fold dilutions of the 5-25 ⁇ g/ml GA RS samples and of the test samples; and c) 15-20 fold dilution of the positive control sample
  • the ELISA protocol for measuring IL-2 levels was performed according to the manufacturer's recommendations. If the optical density of any sample reached the upper/lower limits of the plate reader, the sample was re-analyzed at a higher/lower dilution, respectively.
  • the mean absorbance was subtracted of the blank sample (zero IL-2 standard point) from the absorbance of standards, samples and controls and calculated for each set of triplicate the mean
  • the suspected outlier was designated X x . All other measurements were labeled in reference to the suspected outlier, e.g., X 2 was the value next to the suspected outlier, X 3 was second value from the suspected outlier, X k was the farthest from the suspected outlier and X ⁇ was the value second from the farthest, etc.
  • the absorbance of each of the blank samples was ⁇ 10% of the mean absorbance of the highest concentration of the IL-2 standard. If one of the blank replicates was beyond the above limits, it was rejected and duplicate samples were used.
  • the IL-2 standard curve was graphed according to the manufacturer's recommendations. IL-2 standards that exhibit poor sensitivity or. sample processing error were able to be rejected if a minimum of six non-zero concentration IL-2 standards remained in the curve.
  • the back-calculated standard concentration had a relative error (RE) greater than 20% for the lower calibration point and ⁇ 15% for all other concentrations.
  • the IL-2 calibration curve was constructed from at least six non-zero concentration points (at least 17 calibration points) , covering the range of expected concentrations. The standard curve range was able to be truncated if the high or low concentrations failed.
  • the R 2 of the linear regression curve was ⁇ 0.97.
  • the concentration of IL-2 was calculated in all samples from the linear regression plot of the IL-2 standard, utilizing the equation of the linear regression curve. The final concentration of IL-2 was calculated in all samples by multiplying by the samples' dilution factor.
  • Negative Control (MBP Peptide)
  • the final concentration of IL-2 in at least 2 out of the 3 replicates of the negative control sample was below the levels of IL-2 measured for the lowest calibration point of the GA RS curve .
  • the final concentration of IL-2 in at least 2 out of the 3 replicates of the positive control sample was similar to or above the level of IL-2 in the highest calibration point of the GA RS curve.
  • the GA RS curve was plotted on a log-log scale, with log IL-2 concentration on the y-axis and log GA RS concentration on the x-axis.
  • the calibration curve was constructed from at least five non- zero concentrations (at least 14 calibration points) . Calibration points were rejected as described for the IL-2 standard points. The best- fit regression curve was computed through the standard points.
  • the R 2 was ⁇ 0.97.
  • the slope ( ⁇ ) was ⁇ 0.77.
  • the dose-response curve of each test sample was plotted on a log-log scale, with log IL-2 concentration on the y-axis and log GA DS concentration on the x-axis.
  • the best fit regression curve was computed through the sample points.
  • the slope ( ⁇ *) was within the following range: ⁇ x 0.635 ⁇ ⁇ * ⁇ ⁇ x 1.365.
  • the components 3 and 4 were included in the random error term due to non-significant deviations from linearity and parallelism, respectively.
  • the total sum of squares was partitioned into 3 components (SS-Regression, SS-Preparation and SS-Error) , the appropriate number of degrees of freedom and the F-test for significance .
  • the % potency of the tested batch was calculated and the 95% fiducial limits for the estimated potency as described below:
  • Step 1 Compute the transformation of the given data (GA. Batch and GA. RS. ) into log 1 ⁇ scale :
  • Step 2 Calculate the common slope for the linear regression based on all measured data points via. the formula:
  • Y is an overall mean value of log 10 ( response);
  • X is an overall mean value of log 10 (dose).
  • Step 3 Calculate the sum of squares due to regression on log 10 (dose) aa:
  • Step A Calculate the random error sum of squares aa:
  • YK , Xu are mean log 10 (response) and log 0 (dose) values, respectively, of preparation k.
  • ⁇ Step 5 Calculate the Mean Square Error term as following: DF E R R (random error degrees of freedom) »N-3;
  • Step 7 Calculate the point estimate of the relative potency aa following:
  • Step 8 Calculate the expression denoted by C via the formula:
  • Step 9 Calculate the logarithms of lower and upper limits of 95% Fiducial Interval:
  • Step O Transform the values computed in the previous step into original scale by taking of anti-logarithms of the resulting log-limits and multiply by 100%.
  • the estimated potency of GA DS batch was not less than 80% and not more than 125% of the stated potency.
  • the LN cell count and ELISA plates template were recorded.
  • the original ELISA reader records and the result form were filed.
  • the LN cells were derived from female (SJL x BALB/C) Fl mice immunized with 250 ⁇ g GA RS in CFA 9-11 days earlier were cultured in the presence of various concentrations of GA RS. The cells were incubated with GA RS for 18-24 hours at 37°C in a 5% C0 2 humidified incubator. Subsequently, the cultures were centrifuged and the supernatants collected and assayed for cytokines by ELISA.
  • the ELISA was performed using biotinylated antibodies specific to the cytokine and strepavidin-horseradish peroxidase (HRP) conjugated for detection. Each plate ran included blank control
  • QC quality control
  • Table 4 the maximal levels measured for each cytokine are presented in arbitrary units: (-) ⁇ detection limit; (+) up to - 400 pg/ml; and (++) > 400 pg/ml .
  • Table 4 shows that in response to GA RS in culture, the LN cells secreted IL-2, INF- ⁇ , IL-10 and IL-6, while TNF-o:, IL-4, IL-13 and TGF- ⁇ were not detected in the culture media.
  • the first experiment tested the effect of GA RS (immunizing antigen) dose on T-cell responses in the LN and in the spleen.
  • Two groups of 10 mice each were immunized with either 250 ⁇ g GA in CFA (group 1) (as in the EAE blocking test) or with 10 ⁇ g GA in CFA (group 2) .
  • Primary cultures were prepared from both the LN and the spleens of the immunized mice. The cultures were incubated overnight with various doses of GA RS and afterwards the culture media were collected and assayed for IL-2 as in Experiment 2A. The results in Fig.
  • one group was injected with 250 ⁇ g GA RS in CFA and the second group with 10 mg GA in ICFA.
  • the dose of GA RS in the second group was higher since ICFA, a weaker adjuvant, was used.
  • the response of the LN cells from both groups to GA RS was tested in vi tro .
  • Fig. 5 shows that immunization with 250 ⁇ g GA RS in CFA induced a much stronger response in culture, although a much lower dose of antigen was used.
  • Fig. 6 shows the response of the LN cells to GA RS in culture, prepared 9, 10 and 11 days following immunization. Since the dose-response of IL-2 secretion was similar on all days, the immunization period may last for 9-11 days.
  • mice lymphoid cells are usually maintained in RPMI medium, supplemented with 1% normal mouse serum.
  • Normal mouse serum may contain endogenous IL-2 that can be detected by the anti mouse IL-2 monoclonal antibodies used in the ELISA kit.
  • the use of different lots of normal serum may increase the inter-day variations of the in vi tro test.
  • Fig. 7 shows the results of a representative experiment that compares the response of LN cells to GA RS in different culture media. The best responses were observed when serum- free media were used.
  • the dose-response range was left-shifted in the absence of serum. This can be explained by previous studies showing that GA binds to albumin and to other serum proteins. This binding may reduce the availability of GA in culture to interactions with APCs, and thus higher concentrations of GA are required to stimulate the T cells. Based on these results, the optimum medium seems to be DCCM-1 medium.
  • IL-2 is an autocrine and paracrine growth factor that is essential for clonal T-cell proliferation and for functional properties of B cells and macrophages. Following stimulatibn of the culture with GA RS, IL-2 is secreted by the activated
  • Fig. 8 shows that the peak of IL-2 levels in the culture media is between 18-21 hours.
  • the reduction in IL-2 levels in samples collected from 24-48 hours can be explained by the consumption of IL-2 by the LN cells.
  • the optimum time for supernatant collection appears to be after 18-21 hours of incubation.
  • the method relies on accurate measurements of IL-2 in samples of GA RS and test samples.
  • the levels of IL-2 were measured by OptEIA (Pharmingen, Cat. # 2614KI) - an ELISA kit specific for mouse IL-2. This ELISA kit is very sensitive and the results are accurate and reproducible.
  • the culture media were collected and kept at -20°C before being analyzed by the ELISA.
  • Preliminary studies of the stability of IL-2 in culture media show that the cytokine is stable for one week at -20°C (Fig. 9) . Therefore, the culture media of the in-vitro test samples can be kept at -20°C for up to one week prior to measuring IL-2.
  • the results of this experiment also demonstrated that the ELISA results are very reproducible -- the levels of IL-2 measured in the samples were practically identical in two ELISA plate runs performed on two different days one week apart.
  • the statistical validation was carried out based on GA RS calibration curves calculated and evaluated separately for each one out of 21 plates received for the analysis. These 21 samples were gathered at different times over an approximately four-month period. The GA concentration range for the given plates varied from 0.25 to 50 ⁇ g/ml. The following validation characteristics derived from the GA RS calibration curves constituted the main concern of the analysis:
  • the response variable was a log-transformed mean of the 3 replicates measured at each calibration point. This model was fitted to each calibration sample and the appropriate statistics (R 2 , intercept, and slope) were calculated for each fitted curve. The value of R 2 reflected the ratio of the residual sum of squares (RSS) to the total sum of squares (TSS) via the formula:
  • R 2 1-RSS/TSS
  • the linear range was determined based on the following criteria:
  • the basic measure of (in) accuracy used was the percent difference between the mean of the estimates of concentration and the true concentration in the triplicate samples:
  • the goal of the analysis was to propose acceptance criteria for the fitted calibration curve which ensured that the accuracy and precision of the method were adequate.
  • the acceptance criteria were based on the R 2 and the slope of the GA RS calibration curve. About 80% of the plates could be characterized by small inaccuracy values ( ⁇ 13%) and by good precision (1.1% - 6.7%) . For these 16 "well behaved" standard curves, the following results were obtained:
  • the mean ( ⁇ SD) accuracy value for the method was: 8.0% + 2.3%;
  • Y low and i gh were the lowest and highest log (response) values permitted by the highest allowable (in) accuracy of ⁇ 12.6%.
  • Y s , Y ⁇ , X s , X ⁇ were the mean log (responses) and log (doses) of the standard and test preparations, respectively.
  • B - was a common slope for the standard and test log (dose)- log (response) lines.
  • the least- squares estimate of the common slope - B was a weighted average of the least-squares estimates of two slopes separately from the standard line and the test line. Taking the anti-logarithm of the expression above, one was able to obtain a point estimate of the "true% potency" of a test preparation relatively to its standard:
  • the batch analysis data was obtained from different experiments performed on different days by different operators. Validation trials of the standard procedure of Example 1 were carried out by a series of experiments, each involving: 1) immunization of mice with 250 ⁇ g GA RS in CFA; 2) preparation of a primary culture from the LN cells 9-11 days following immunization; 3) incubation of the LN cells with various concentrations of GA RS and with test samples; 4) collection of the culture media and analysis of IL-2 levels by ELISA; 5) plotting a GA RS curve based on triplicate IL-2 measurements performed at 6 dose levels from 1-25 ⁇ g/ml; and 6) comparison of the T cell response to each test sample to the response to the RS batch (in triplicate) at two concentrations within the linear range (5 and 10 ⁇ g/ml) .
  • the % CV was also calculated for each triplicate in order to detect any problems associated with variability between triplicates (normally, the % CV between triplicates should not exceed 10- 15%) . For the given data,
  • the validation characteristics used to provide an overall knowledge of the capabilities of the analytical procedure were: linearity, range, accuracy, precision, specificity and robustness.
  • the validation criteria and analyses were based on the ICH consensus guideline, "Validation of Analytical Procedures: Methodology", November 1996 (CPMP/ICH/281/95) .
  • Statistical methods recommended in "European Pharmacopoeia" guideline were adapted to the given data for analysis purposes.
  • a dose-response curve of GA RS batch was used to calculate the relative response of the cells to the tested samples.
  • Each calibration curve included at least five points (without zero) . Twenty-one calibration curves collected from different in vi tro tests, performed during the development and the validation stages, were plotted and evaluated for each plate .
  • the GA RS curve should be comprised of at least 6 calibration points, one with zero concentration (negative control) and at least 5 concentrations of GA RS in the range between 1 and 25 ⁇ g/ml.
  • Linear regression of log 10 (IL-2 concentration) versus log 10 (GA RS concentration) should have an R 2 ⁇ 0.97 and a slope ⁇ 0.77.
  • the accuracy of the method was established across the specified linear range of the GA RS curve. Statistical analysis of the data revealed that the mean accuracy of the method was: 8.0% ⁇ 2.3%.
  • the GA specific T cell response to a GA DS batch was measured repeatedly, 3 times, in the same in vi tro test. Three weights of the same batch were each diluted to 5 and 10 ⁇ g/ml and incubated with the GA-specific T-cells. The levels of IL-2 in the culture media of the test samples and of the GA RS samples, were measured by ELISA in triplicate. The % potency and 95% fiducial limits of the cells to each replicate were calculated relative to the GA RS. Table 6 shows the % response calculated for each replicate.
  • the % response of a GA DS batch was tested in 3 different in vi tro tests, performed in different days, by 3 different investigators from the same laboratory.
  • Table 7 summarizes the % potency and 95% fiducial limits determined for this batch in the 3 repeated experiments.
  • the discrimination of the method was tested at 3 levels: 1) discrimination between samples incubated with/without GA RS (matrix effect) ; 2) discrimination between GA RS and other related and non-related proteins and peptides, including GA DS; and 3) discrimination between GA RS and GA related copolymers in which the peptide sequences have been deliberately modified.
  • GA-specific T cells were induced by immunization of female (SJL x BALB/C) Fl mice mouse with 250 g GA RS in CFA. This dose of GA is routinely used for testing the biological activity of GA batches in this mouse strain using the EAE blocking test .
  • the control group in this experiment was injected with CFA alone.
  • LN cells were removed from the both groups of mice. The cells were incubated with GA RS for 18-24 hours at 37°C in a 5% C0 2 humidified incubator.
  • IL-2 interleukin-2
  • HRP strepavidin-horseradish peroxidase conjugate for detection
  • Each in vi tro test included a positive control (Con A, a non-specific T-cell stimulant) and a negative control (no GA or any other antigen) .
  • Fig. 2 shows that the LN cells from mice immunized with GA RS secrete IL-2 dose dependently in response to GA RS in culture, while LN cells from the control mice do not respond to GA. RS in culture.
  • the levels of IL-2 in the negative control samples is usually below or close to the ELISA detection limit (approximately 3 pg/ml) . These IL-2 levels are always below the levels secreted by the lowest calibration point of GA RS (1 ⁇ g/ml) .
  • Table 9 shows that in this experimental system the GA-specific T cells did not respond to either human MBP (myelin basic protein) , the MBP immunodominant peptide pp.87-99 (an encephalitogenic peptide), or its analog pp .87-99 ⁇ 96 (an EAE suppressor peptide) .
  • Lysozyme a non-relevant basic protein, was also not recognized by the GA-specific T cells.
  • TN- 35 and TV-109 were peptides with a molecular weight of 3757 and 11727, respectively (PCT International Publication No. WO 00/18794) .
  • IL-2 was measured as in Experiment 3G(i) .
  • Fig. 12A shows that the LN cells cross-reacted with both standard batches.
  • the dose-response curves of IL-2 secretion (measured by ELISA as above) by both batches were similar, indicating that the tested batches shared similar T-cell epitopes.
  • Comparison between GA RS and a Copaxone ® batch shows that the GA RS-specific T cells also cross-reacted with the DP batch, and that mannitol, the excipient in the Copaxone ® formulation, did not affect or interfere with the T-cell responses (Fig. 12B) .
  • this method provides an indication of batch-to-batch reproducibility.
  • Table 10 shows that the GA-specific T-cells did not respond to the 3 amino acid copolymers lacking lysine, alanine or tyrosine.
  • the % response of the cells to the batch XX was relatively high and out of the method specifications (100 ⁇ 30%) , indicating that the method might be sensitive to modifications in the production process.
  • the high % response can also be explained by the sensitivity of the test to the MW of GA peptides, as demonstrated .
  • the consistency and robustness of the defined acceptance criteria was examined by comparing the resulting estimates of the relative potency obtained for the repeated GA batches .
  • the batch analysis data included a number of repeated GA batches . Two batches were measured on three different days by different operators. One batch was tested on two different days by different operators, as well.
  • the acceptance criteria were determined to be :
  • the estimated relative potency was not less than 80% and not more than 125% of the standard potency; and 3.
  • the 95% Fiducial Limits of error of the estimated relative potency were not less than 70% and not more than 143% of the standard potency.
  • the method was very specific to GA peptides and is sensitive to the average MW of the peptide mixture.
  • MBP was not recognized by the GA-specific T cells.
  • MBP immunodominant peptides both encephalitogenic and suppressive peptides
  • Single peptides with amino-acid composition similar to that of GA did not stimulate the T cells.
  • Critical parameters in the immunization procedure, as well as in the in-vitro reaction, were optimized during this experiment. This experiment showed that the method was very reproducible and robust .
  • the method can be adapted to standardize other T cell antigens for use in pharmaceutical compositions.
  • a primary culture of T cells specific to an antigen RS instead of GA RS, can be made from animals immunized against the antigen RS .
  • the cytokine production of this culture in response to antigen RS and in response to the sample antigen can be measured.
  • the cytokine production in response to antigen RS can be plotted against the concentration of antigen RS to create a standard curve.
  • the cytokine production in response to the sample antigen can be compared to the standard curve to determine whether the antigen is within the acceptable range of potency.
  • the optimum cytokine to monitor can be determined as in Experiment 2A.
  • Conditions for immunization and the in vi tro test may be optimized as in Experiments 2B and C.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • General Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
PCT/US2002/038859 2001-12-04 2002-12-04 Processes for the measurement of the potency of glatiramer acetate WO2003048735A2 (en)

Priority Applications (19)

Application Number Priority Date Filing Date Title
MXPA04005433A MXPA04005433A (es) 2001-12-04 2002-12-04 Proceso para la medicion de la potencia de acetato de glatiramero.
SI200230921T SI1459065T1 (sl) 2001-12-04 2002-12-04 Postopki za merjenje potentnosti glatiramer acetata
DK02790028.1T DK1459065T3 (da) 2001-12-04 2002-12-04 Fremgangsmåde til målingen af styrken af glatirameracetat
IL16212702A IL162127A0 (en) 2001-12-04 2002-12-04 Process for the measurement of the potency of glatiramer acetate
EP02790028A EP1459065B1 (en) 2001-12-04 2002-12-04 Processes for the measurement of the potency of glatiramer acetate
DE60237170T DE60237170D1 (de) 2001-12-04 2002-12-04 Verfahren zur messung der wirkstärke von glatirameracetat
NZ533327A NZ533327A (en) 2001-12-04 2002-12-04 Processes for the measurement of the potency of glatiramer acetate
AU2002353059A AU2002353059B2 (en) 2001-12-04 2002-12-04 Processes for the measurement of the potency of glatiramer acetate
CNB028277732A CN1308683C (zh) 2001-12-04 2002-12-04 测量醋酸格拉默功效的方法
JP2003549881A JP4369234B2 (ja) 2001-12-04 2002-12-04 グラチラマー・アセテートの強度の測定のための方法
AT02790028T ATE475883T1 (de) 2001-12-04 2002-12-04 Verfahren zur messung der wirkstärke von glatirameracetat
KR1020047008518A KR100657048B1 (ko) 2001-12-04 2002-12-04 글라티라머 아세테이트의 효능 측정 방법
CA2469393A CA2469393C (en) 2001-12-04 2002-12-04 Processes for the measurement of the potency of glatiramer acetate
IL162127A IL162127A (en) 2001-12-04 2004-05-23 Process for the measurement of the potency of glatiramer acetate
IS7286A IS7286A (is) 2001-12-04 2004-05-26 Aðferðir til þess að mæla hæfi glatiramerasetats
ZA2004/04472A ZA200404472B (en) 2001-12-04 2004-06-07 Processes for the measurement of the potency of glatiramer acetate
NO20042816A NO338247B1 (no) 2001-12-04 2004-07-02 Fremgangsmåter for måling av kraften av glatirameracetat
HK05102405.0A HK1070421A1 (en) 2001-12-04 2005-03-19 Processes for the measurement of the potency of glatiramer acetate
IL198285A IL198285A (en) 2001-12-04 2009-04-22 A process for measuring the potency of Galtirmer @ acetate

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US33876701P 2001-12-04 2001-12-04
US60/338,767 2001-12-04

Publications (2)

Publication Number Publication Date
WO2003048735A2 true WO2003048735A2 (en) 2003-06-12
WO2003048735A3 WO2003048735A3 (en) 2003-09-18

Family

ID=23326090

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/038859 WO2003048735A2 (en) 2001-12-04 2002-12-04 Processes for the measurement of the potency of glatiramer acetate

Country Status (22)

Country Link
US (6) US7429374B2 (zh)
EP (1) EP1459065B1 (zh)
JP (1) JP4369234B2 (zh)
KR (1) KR100657048B1 (zh)
CN (1) CN1308683C (zh)
AT (1) ATE475883T1 (zh)
AU (1) AU2002353059B2 (zh)
CA (1) CA2469393C (zh)
CY (1) CY1110878T1 (zh)
DE (1) DE60237170D1 (zh)
DK (1) DK1459065T3 (zh)
ES (1) ES2349033T3 (zh)
HK (1) HK1070421A1 (zh)
IL (3) IL162127A0 (zh)
IS (1) IS7286A (zh)
MX (1) MXPA04005433A (zh)
NO (1) NO338247B1 (zh)
NZ (1) NZ533327A (zh)
PT (1) PT1459065E (zh)
SI (1) SI1459065T1 (zh)
WO (1) WO2003048735A2 (zh)
ZA (1) ZA200404472B (zh)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7022663B2 (en) 2000-02-18 2006-04-04 Yeda Research And Development Co., Ltd. Oral, nasal and pulmonary dosage formulations of copolymer 1
US7074580B2 (en) 1998-09-25 2006-07-11 Yeda Research And Development Co., Ltd. Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
US7279172B2 (en) 1998-07-23 2007-10-09 Yeda Research And Development Co., Ltd. Treatment of autoimmune conditions with copolymer 1 and related copolymers
US7425332B2 (en) 1998-07-23 2008-09-16 Yeda Research And Development Co., Ltd. Treatment of autoimmune conditions with Copolymer 1 and related Copolymers
US7429374B2 (en) 2001-12-04 2008-09-30 Teva Pharmaceutical Industries, Ltd. Process for the measurement of the potency of glatiramer acetate
US7560100B2 (en) 2004-09-09 2009-07-14 Yeda Research And Development Co., Ltd. Mixtures of polypeptides, compositions containing and processes for preparing same, for treating neurodegenerative diseases
US7884187B2 (en) 2008-04-16 2011-02-08 Momenta Pharmaceuticals, Inc. Analysis of amino acid copolymer compositions
EP2381254A1 (en) * 2007-06-21 2011-10-26 Momenta Pharmaceuticals, Inc. Copolymer assay
US8324348B1 (en) 2011-07-11 2012-12-04 Momenta Pharmaceuticals, Inc. Evaluation of copolymer diethylamide
EP2642290A1 (en) 2012-03-19 2013-09-25 Synthon BV Glatiramer acetate human monocytic cell line-based potency assay
WO2013139728A1 (en) 2012-03-19 2013-09-26 Synthon Bv Glatiramer acetate human monocyte cell-based potency assay
US8709433B2 (en) 2010-10-11 2014-04-29 Teva Pharmaceutical Industries Ltd. Cytokine biomarkers as predictive biomarkers of clinical response for Glatiramer acetate
US8759302B2 (en) 2010-03-16 2014-06-24 Teva Pharmaceutical Industries, Ltd. Methods of treating a subject afflicted with an autoimmune disease using predictive biomarkers of clinical response to glatiramer acetate therapy in multiple sclerosis
US8815511B2 (en) 2011-10-10 2014-08-26 Teva Pharmaceutical Industries, Ltd. Determination of single nucleotide polymorphisms useful to predict response for glatiramer acetate
WO2014174070A1 (en) 2013-04-26 2014-10-30 Synthon B.V. Glatiramer acetate human monocytic cell line-based potency assay
US9155776B2 (en) 2009-08-20 2015-10-13 Yeda Research & Development Co., Ltd. Low frequency glatiramer acetate therapy
US9155775B1 (en) 2015-01-28 2015-10-13 Teva Pharmaceutical Industries, Ltd. Process for manufacturing glatiramer acetate product
WO2016128957A1 (en) * 2015-02-12 2016-08-18 Insight Biopharmaceuticals Ltd. Methods of determining relative potency of glatiramer acetate and use thereof in preparing a batch of glatiramer acetate as acceptable for pharmaceutical use
EP2941274A4 (en) * 2013-01-04 2016-11-16 Teva Pharma CHARACTERIZATION OF A MEDICINAL PRODUCT ASSOCIATED WITH GLATIRAMÈRE ACETATE
EP2971173A4 (en) * 2013-03-14 2016-11-23 Mylan Inc GLATIRAMERATE ACETATE POWER TEST BY MRM RESPONSE BIOMARKERS
US9617596B2 (en) 2012-10-10 2017-04-11 Teva Pharmaceutical Industries, Ltd. Biomarkers predictive for clinical response for glatiramer acetate
US9702007B2 (en) 2013-10-21 2017-07-11 Teva Pharmaceuticals Industries, Ltd. Genetic markers predictive of response to glatiramer acetate
US9995734B2 (en) 2013-10-24 2018-06-12 Mylan Inc. Human T cell line assay for evaluating the immunologic identity of glatiramer acetate preparations
US11167003B2 (en) 2017-03-26 2021-11-09 Mapi Pharma Ltd. Methods for suppressing or alleviating primary or secondary progressive multiple sclerosis (PPMS or SPMS) using sustained release glatiramer depot systems
USRE49251E1 (en) 2010-01-04 2022-10-18 Mapi Pharma Ltd. Depot systems comprising glatiramer or pharmacologically acceptable salt thereof

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020077278A1 (en) 2000-06-05 2002-06-20 Yong V. Wee Use of glatiramer acetate (copolymer 1) in the treatment of central nervous system disorders
WO2002076503A1 (en) * 2000-06-20 2002-10-03 Mayo Foundation For Medical Education And Research Treatment of central nervous system diseases by antibodies against glatiramer acetate
US20070244056A1 (en) * 2004-03-03 2007-10-18 Liat Hayardeny Combination Therapy With Glatiramer Acetate and Riluzole
US7495072B2 (en) * 2004-09-09 2009-02-24 Teva Pharmaceutical Industries, Ltd. Process for preparation of mixtures of polypeptides using purified hydrobromic acid
US8324641B2 (en) * 2007-06-29 2012-12-04 Ledengin, Inc. Matrix material including an embedded dispersion of beads for a light-emitting device
CA2588908C (en) * 2004-11-29 2014-09-23 Yeda Research And Development Co. Ltd Induction of neurogenesis and stem cell therapy in combination with copolymer 1
US20060172942A1 (en) * 2005-02-02 2006-08-03 Teva Pharmaceutical Industries, Ltd. Process for producing polypeptide mixtures using hydrogenolysis
US20080261894A1 (en) * 2005-02-17 2008-10-23 Rivka Kreitman Combination Therapy with Glatiramer Acetate and Rasagiline for the Treatment of Multiple Sclerosis
US20060240463A1 (en) * 2005-04-25 2006-10-26 Rappaport Family Institute For Research In The Medical Sciences Markers associated with the therapeutic efficacy of glatiramer acetate
US20090149541A1 (en) * 2007-11-28 2009-06-11 Yafit Stark Method of delaying the onset of clinically definite multiple sclerosis
JP2013511724A (ja) * 2009-11-17 2013-04-04 アレス トレーディング ソシエテ アノニム 血清タンパク質に基づく指向性配列ポリマー組成物の検出による、指向性配列ポリマー組成物の設計、生物学的利用性、及び有効性を改良するための方法
US8575198B1 (en) 2011-09-07 2013-11-05 Momenta Pharmaceuticals, Inc. In-process control for the manufacture of glatiramer acetate
US20130210054A1 (en) * 2012-02-09 2013-08-15 Momenta Pharmaceuticals, Inc. Amino Acid Copolymer Assay

Family Cites Families (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL36670A (en) * 1971-04-21 1974-09-10 Sela M Therapeutic basic copolymers of amino acids
US3991210A (en) * 1974-03-11 1976-11-09 The Dow Chemical Company Acetamidine urinary antiseptics
US4129666A (en) * 1977-04-29 1978-12-12 Walter Wizerkaniuk Method of providing pellets with a water insoluble coating using a melt
US4339431A (en) * 1980-12-31 1982-07-13 Colgate-Palmolive Company Anticalculus oral composition
JPS6053535A (ja) 1983-09-02 1985-03-27 Nitto Boseki Co Ltd 規則性ポリアミノ酸樹脂の製造方法
SU1182051A1 (ru) 1984-04-28 1985-09-30 Таджикский государственный университет им.В.И.Ленина Политрипептиды,обладающие энантовой селективностью в реакци х гидролиза @ -нитрофениловых эфиров карбобензокси - @ - и @ -аланина
EP0228448B1 (en) 1985-06-18 1991-08-28 Emory University Use of biologically active copolymers for the manufacture of a medicament for stimulating the growth of an animal
DD257174A3 (de) 1985-12-20 1988-06-08 Ve Forschungszentrum Biotechno Verfahren zur herstellung der peptide z-ala-ala-lon-p-nitranilid oder z-asp-phe-ome mit metalloprotease
SU1469826A1 (ru) 1986-05-30 1995-11-20 Институт Высокомолекулярных Соединений Ан Ссср Сополимер l-лизина с l-глутаминовой кислотой, содержащий дофаминовые боковые группы, обладающий пролонгированной гипотензивной активностью и компенсаторным эффектом при геморрагическом шоке, и способ его получения
US5554372A (en) * 1986-09-22 1996-09-10 Emory University Methods and vaccines comprising surface-active copolymers
ATE258065T1 (de) 1987-06-24 2004-02-15 Brigham & Womens Hospital Behandlung von autoimmun-erkrankungen durch orale verabreichung von autoantigenen
CA2009996A1 (en) 1989-02-17 1990-08-17 Kathleen S. Cook Process for making genes encoding random polymers of amino acids
SU1690368A1 (ru) 1989-07-20 1995-08-20 Институт Высокомолекулярных Соединений Ан Ссср Статистические сополимеры в качестве низкотоксичных веществ, обладающих пролонгированным гипотензивным действием, и способ их получения
DE3930733A1 (de) 1989-09-14 1991-03-28 Roehm Gmbh Verfahren zur herstellung eines komplexierten arzneimittels
FR2658076B1 (fr) * 1990-02-12 1992-06-12 Sanofi Sa Composition cosmetique contenant des copolymeres d'aminoacides, utile comme agent hydratant.
AU8305491A (en) 1990-08-01 1992-03-02 Cytel Corporation Novel immunosuppressant peptides
US5668117A (en) * 1991-02-22 1997-09-16 Shapiro; Howard K. Methods of treating neurological diseases and etiologically related symptomology using carbonyl trapping agents in combination with previously known medicaments
US5734023A (en) * 1991-11-19 1998-03-31 Anergen Inc. MHC class II β chain/peptide complexes useful in ameliorating deleterious immune responses
US5583031A (en) * 1992-02-06 1996-12-10 President And Fellows Of Harvard College Empty major histocompatibility class II heterodimers
WO1994003484A1 (en) * 1992-07-31 1994-02-17 Merrell Dow Pharmaceuticals Inc. Synthetic peptide lung surfactants having covalently bonded antioxidants
WO1994026774A1 (en) 1993-05-19 1994-11-24 Cytel Corporation Novel treatments for allergic diseases
AU703451B2 (en) * 1994-03-31 1999-03-25 Vertex Pharmaceuticals Incorporated N-(pyrimidinyl)-aspartic acid analogs as interleukin-1beta converting enzyme inhibitors
AU2238395A (en) 1994-04-01 1995-10-23 Immulogic Pharmaceutical Corporation Haptenated peptides and uses thereof
US5591629A (en) * 1994-04-29 1997-01-07 Mayo Foundation For Medical Education & Research Monoclonal antibodies which promote central nervous system remyelination
US5502022A (en) * 1994-05-16 1996-03-26 Biosepra, Inc. Chromatography adsorbents utilizing mercapto heterocyclic ligands
AU7242994A (en) 1994-05-20 1995-12-18 United States Of America, As Represented By The Secretary Of The Army, The Model for testing immunogenicity of peptides
US5800808A (en) * 1994-05-24 1998-09-01 Veda Research And Development Co., Ltd. Copolymer-1 improvements in compositions of copolymers
GB9411292D0 (en) 1994-06-06 1994-07-27 Teva Pharma Pharmaceuticals compositions
US5858964A (en) * 1995-04-14 1999-01-12 Yeda Research And Development Co. Ltd. Pharmaceutical compositions comprising synthetic peptide copolymer for prevention of GVHD
US5627206A (en) * 1995-06-02 1997-05-06 Warner-Lambert Company Tricyclic inhibitor of matrix metalloproteinases
US5665764A (en) * 1995-06-02 1997-09-09 Warner-Lambert Company Tricyclic inhibitors of matrix metalloproteinases
US5719296A (en) 1995-10-30 1998-02-17 Merck & Co., Inc. Pseudopeptide lactam inhibitors of peptide binding to MHC class II proteins
DE69718968T2 (de) 1996-01-17 2003-10-09 Taiho Pharmaceutical Co Ltd 3-(bis-substituierte-phenylmethylen)oxindol-derivate
US5716946A (en) * 1996-02-13 1998-02-10 Wisconsin Alumni Research Foundation Multiple sclerosis treatment
IL119989A0 (en) 1997-01-10 1997-04-15 Yeda Res & Dev Pharmaceutical compositions for oral treatment of multiple sclerosis
US6214791B1 (en) * 1997-01-10 2001-04-10 Yeda Research And Development Co. Ltd. Treatment of multiple sclerosis through ingestion or inhalation of copolymer-1
US6024981A (en) * 1997-04-16 2000-02-15 Cima Labs Inc. Rapidly dissolving robust dosage form
US6114317A (en) * 1998-05-21 2000-09-05 Wisconsin Alumni Research Foundation Method of locking 1α-OH of vitamin D compounds in axial orientation
AU766498B2 (en) 1998-07-23 2003-10-16 President And Fellows Of Harvard College, The Synthetic peptides and methods of use for autoimmune disease therapies
ES2527760T3 (es) * 1998-07-23 2015-01-29 Yeda Research And Development Co., Ltd. Tratamiento de enfermedad de Crohn con copolímero 1 y polipéptidos
IL141021A0 (en) 1998-07-23 2002-02-10 Yeda Res & Dev Treatment of autoimmune conditions with copolymer 1 and related copolymers
DK2239269T3 (da) 1998-09-25 2013-04-29 Yeda Res & Dev Anvendelse af peptider afledt af copolymer-1 som molekylvægtsmarkører
US6800287B2 (en) * 1998-09-25 2004-10-05 Yeda Research And Development Co., Ltd. Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
US6514938B1 (en) * 1998-09-25 2003-02-04 Yeda Research And Development Co. Ltd. At The Weizmann Institute Of Science Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
AU6281599A (en) 1998-10-02 2000-04-26 Yeda Research And Development Co. Ltd. Alternate day administration of copolymer 1 for treating autoimmune diseases
EP1128839A4 (en) 1998-11-12 2002-10-09 Yeda Res & Dev PHARMACEUTICAL COMPOSITIONS CONTAINING COPOLYMERS OF SYNTHETIC PEPTIDES AND METHODS FOR PREVENTING AND TREATING GVHD AND HVGD
AU780188B2 (en) * 2000-01-20 2005-03-03 Yeda Research And Development Co. Ltd. The use of copolymer 1 and related peptides and polypeptides and T cells treated therewith for neuroprotective therapy
US7022663B2 (en) * 2000-02-18 2006-04-04 Yeda Research And Development Co., Ltd. Oral, nasal and pulmonary dosage formulations of copolymer 1
CN1221281C (zh) 2000-02-18 2005-10-05 耶达研究与开发有限公司 共聚物1的口服、鼻部和肺部用剂型
CA2409515C (en) 2000-05-10 2015-11-24 Mayo Foundation For Medical Education & Research Human igm antibodies with the capability of inducing remyelination, and diagnostic and therapeutic uses thereof particularly in the central nervous system
US20020107388A1 (en) * 2000-05-12 2002-08-08 Vandenbark Arthur A. Methods of identifying and monitoring disease-associated T cells
AU7528001A (en) 2000-06-05 2001-12-17 Teva Pharma The use of glatiramer acetate (copolymer 1) in the treatment of central nervous system disorders
US20020077278A1 (en) * 2000-06-05 2002-06-20 Yong V. Wee Use of glatiramer acetate (copolymer 1) in the treatment of central nervous system disorders
JP4328090B2 (ja) * 2000-06-07 2009-09-09 イエダ リサーチ アンド デベロップメント カンパニー リミテッド 神経細胞の保護療法におけるコポリマー1、関連ペプチドおよびポリペプチドならびにそれらによって処理されたt細胞の使用
WO2002076503A1 (en) 2000-06-20 2002-10-03 Mayo Foundation For Medical Education And Research Treatment of central nervous system diseases by antibodies against glatiramer acetate
CN2469393Y (zh) 2001-03-08 2002-01-02 祁新 暖气热水器
CN1152258C (zh) * 2001-03-19 2004-06-02 上海肿瘤特殊项目检测中心 一种检测化学药物在体内抗癌功效的方法
US6855270B2 (en) * 2001-06-07 2005-02-15 The University Of Kentucky Research Foundation Nanoscintillation systems for aqueous-based liquid scintillation counting
CA2469393C (en) 2001-12-04 2010-05-25 Teva Pharmaceutical Industries, Ltd. Processes for the measurement of the potency of glatiramer acetate
EP1565486A2 (en) 2002-11-13 2005-08-24 Apotex Pharmachem Inc. Process for the preparation of glatiramer acetate by polymerisation of n-carboxy anhydrides of l-alanine, l-tyrosine, benzyl l-glutamate and benzyloxycarbonyl l-lysine
EP1592384B1 (en) * 2003-01-21 2012-10-31 Yeda Research And Development Co., Ltd. Cop 1 for treatment of inflammatory bowel diseases
SI1638589T1 (sl) * 2003-05-14 2014-07-31 Teva Pharmaceutical Industries Ltd. Kombinirana terapija z glatiramer acetatom in mitoksantronom za zdravljenje multiple skleroze
WO2005028630A2 (en) * 2003-09-19 2005-03-31 Sangamo Biosciences, Inc. Engineered zinc finger proteins for regulation of gene expression
US20050170004A1 (en) 2003-10-31 2005-08-04 Vered Rosenberger Nanoparticles for drug delivery
US7560100B2 (en) * 2004-09-09 2009-07-14 Yeda Research And Development Co., Ltd. Mixtures of polypeptides, compositions containing and processes for preparing same, for treating neurodegenerative diseases
US7495072B2 (en) * 2004-09-09 2009-02-24 Teva Pharmaceutical Industries, Ltd. Process for preparation of mixtures of polypeptides using purified hydrobromic acid
ATE440888T1 (de) 2004-10-29 2009-09-15 Sandoz Ag Verfahren zur herstellung von glatiramer
US20060172942A1 (en) * 2005-02-02 2006-08-03 Teva Pharmaceutical Industries, Ltd. Process for producing polypeptide mixtures using hydrogenolysis
US20060240463A1 (en) * 2005-04-25 2006-10-26 Rappaport Family Institute For Research In The Medical Sciences Markers associated with the therapeutic efficacy of glatiramer acetate
WO2007030573A2 (en) * 2005-09-09 2007-03-15 Yeda Research And Development Co. Ltd. Polypeptides useful for molecular weight determinations
EP2046817B1 (en) 2006-07-05 2009-12-16 Momenta Pharmaceuticals, Inc. Improved process for the preparation of copolymer-1
ITRM20070552A1 (it) 2007-10-23 2009-04-24 Acqua Minerale S Benedetto S P Contenitore in materiale plastico

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
AHARONI ET AL: 'Bystander suppression of experimental autoimmune encephalomyelitis by T cell lines and clones of the Th2 type induced by copolymer 1' JOURNAL OF NEUROIMMUNOLOGY vol. 91, no. 1-2, November 1998, pages 135 - 146, XP002966941 *
AHARONI ET AL: 'T suppressor hybridoma and interleukin-2 dependent lines induced by copolymer 1 or by spinal cord homogenate down-regulate experimental allergic encephalomyelitis' EUROPEAN JOURNAL OF IMMUNOLOGY vol. 23, 1993, pages 17 - 25, XP002981661 *
DUDA ET AL: 'Human and murine CD4 T cell reactivity to a complex antigen: recognition of the synthesis random polypeptide glatiramer acetate' THE JOURNAL OF IMMUNOLOGY vol. 165, no. 12, 2000, pages 7300 - 7307, XP002966942 *
See also references of EP1459065A2 *
WIESEMANN ET AL: 'Glatiramer acetate (GA) induces IL-13/Il-5 secretion in native T cells' JOURNAL OF NEUROIMMUNOLOGY vol. 119, no. 1, 2001, pages 137 - 144, XP002966943 *

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7279172B2 (en) 1998-07-23 2007-10-09 Yeda Research And Development Co., Ltd. Treatment of autoimmune conditions with copolymer 1 and related copolymers
US7425332B2 (en) 1998-07-23 2008-09-16 Yeda Research And Development Co., Ltd. Treatment of autoimmune conditions with Copolymer 1 and related Copolymers
US7074580B2 (en) 1998-09-25 2006-07-11 Yeda Research And Development Co., Ltd. Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
US7615359B2 (en) 1998-09-25 2009-11-10 Yeda Research And Development Co., Ltd. Copolymer 1 related polypeptides for use as molecular weight markers and for therapeutic use
US7022663B2 (en) 2000-02-18 2006-04-04 Yeda Research And Development Co., Ltd. Oral, nasal and pulmonary dosage formulations of copolymer 1
US7923215B2 (en) 2001-12-04 2011-04-12 Teva Pharmaceutical Industries, Ltd. Process for the measurement of the potency of glatiramer acetate
US7429374B2 (en) 2001-12-04 2008-09-30 Teva Pharmaceutical Industries, Ltd. Process for the measurement of the potency of glatiramer acetate
US7560100B2 (en) 2004-09-09 2009-07-14 Yeda Research And Development Co., Ltd. Mixtures of polypeptides, compositions containing and processes for preparing same, for treating neurodegenerative diseases
US8753833B2 (en) 2007-06-21 2014-06-17 Momenta Pharmaceuticals, Inc. Copolymer assay
EP2381254A1 (en) * 2007-06-21 2011-10-26 Momenta Pharmaceuticals, Inc. Copolymer assay
US8592142B2 (en) 2008-04-16 2013-11-26 Momenta Pharmaceuticals, Inc. Analysis of amino acid copolymer compositions
US8329391B2 (en) 2008-04-16 2012-12-11 Momenta Pharmaceuticals, Inc. Analysis of amino acid copolymer compositions
US10160992B2 (en) 2008-04-16 2018-12-25 Momenta Pharmaceuticals, Inc. Analysis of amino acid copolymer compositions
US7884187B2 (en) 2008-04-16 2011-02-08 Momenta Pharmaceuticals, Inc. Analysis of amino acid copolymer compositions
US9085796B2 (en) 2008-04-16 2015-07-21 Momenta Pharmaceuticals, Inc. Analysis of amino acid copolymer compositions
US9410964B2 (en) 2008-04-16 2016-08-09 Momenta Pharmaceuticals, Inc. Analysis of amino acid copolymer compositions
US9395374B2 (en) 2008-04-16 2016-07-19 Momenta Pharmaceuticals, Inc. Analysis of amino acid copolymer compositions
US9402874B2 (en) 2009-08-20 2016-08-02 Yeda Research & Development Co., Ltd. Low frequency glatiramer acetate therapy
US9155776B2 (en) 2009-08-20 2015-10-13 Yeda Research & Development Co., Ltd. Low frequency glatiramer acetate therapy
USRE49251E1 (en) 2010-01-04 2022-10-18 Mapi Pharma Ltd. Depot systems comprising glatiramer or pharmacologically acceptable salt thereof
US9687522B2 (en) 2010-03-16 2017-06-27 Teva Pharmaceutical Industries, Ltd. Methods of treating a subject afflicted with an autoimmune disease using predictive biomarkers of clinical response to glatiramer acetate therapy in multiple sclerosis
US8759302B2 (en) 2010-03-16 2014-06-24 Teva Pharmaceutical Industries, Ltd. Methods of treating a subject afflicted with an autoimmune disease using predictive biomarkers of clinical response to glatiramer acetate therapy in multiple sclerosis
US9625473B2 (en) 2010-10-11 2017-04-18 Teva Pharmaceutical Industries Ltd. Cytokine biomarkers as predictive biomarkers of clinical response for glatiramer acetate
US9063153B2 (en) 2010-10-11 2015-06-23 Teva Pharmaceuticals Industries Ltd. Cytokine biomarkers as predictive biomarkers of clinical response for glatiramer acetate
US8709433B2 (en) 2010-10-11 2014-04-29 Teva Pharmaceutical Industries Ltd. Cytokine biomarkers as predictive biomarkers of clinical response for Glatiramer acetate
US8324348B1 (en) 2011-07-11 2012-12-04 Momenta Pharmaceuticals, Inc. Evaluation of copolymer diethylamide
US8765911B2 (en) 2011-07-11 2014-07-01 Momenta Pharmaceuticals, Inc. Evaluation of copolymer diethylamide
US8759484B2 (en) 2011-07-11 2014-06-24 Momenta Pharmaceuticals, Inc. Evaluation of copolymer diethylamide
US8815511B2 (en) 2011-10-10 2014-08-26 Teva Pharmaceutical Industries, Ltd. Determination of single nucleotide polymorphisms useful to predict response for glatiramer acetate
EP2642290A1 (en) 2012-03-19 2013-09-25 Synthon BV Glatiramer acetate human monocytic cell line-based potency assay
WO2013139728A1 (en) 2012-03-19 2013-09-26 Synthon Bv Glatiramer acetate human monocyte cell-based potency assay
US9617596B2 (en) 2012-10-10 2017-04-11 Teva Pharmaceutical Industries, Ltd. Biomarkers predictive for clinical response for glatiramer acetate
EP2941274A4 (en) * 2013-01-04 2016-11-16 Teva Pharma CHARACTERIZATION OF A MEDICINAL PRODUCT ASSOCIATED WITH GLATIRAMÈRE ACETATE
US10344330B2 (en) 2013-03-14 2019-07-09 Mylan Inc. Glatiramer acetate response biomarker mRNA potency assay
EP2971173A4 (en) * 2013-03-14 2016-11-23 Mylan Inc GLATIRAMERATE ACETATE POWER TEST BY MRM RESPONSE BIOMARKERS
EP3118626A1 (en) 2013-04-26 2017-01-18 Synhton B.V. Glatiramer acetate human monocytic cell line-based potency assay
EP3118625A1 (en) 2013-04-26 2017-01-18 Synhton B.V. Glatiramer acetate human monocytic cell line-based potency assay
WO2014173463A1 (en) 2013-04-26 2014-10-30 Synthon Bv Glatiramer acetate human monocytic cell line-based potency assay
US9702866B2 (en) 2013-04-26 2017-07-11 Synthon B.V. Glatiramer acetate human monocytic cell line-based potency assay
WO2014174070A1 (en) 2013-04-26 2014-10-30 Synthon B.V. Glatiramer acetate human monocytic cell line-based potency assay
EA031418B1 (ru) * 2013-04-26 2018-12-28 Синтон Б.В. Оценка активности глатирамерацетата на основе линии моноцитарных клеток человека
US9702007B2 (en) 2013-10-21 2017-07-11 Teva Pharmaceuticals Industries, Ltd. Genetic markers predictive of response to glatiramer acetate
US10663457B2 (en) 2013-10-24 2020-05-26 Mylan Inc. Human T cell line assay for evaluating the immunologic identity of glatiramer acetate preparations
US9995734B2 (en) 2013-10-24 2018-06-12 Mylan Inc. Human T cell line assay for evaluating the immunologic identity of glatiramer acetate preparations
US9763993B2 (en) 2015-01-28 2017-09-19 Teva Pharmaceutical Industries Ltd. Process for manufacturing glatiramer acetate product
US9155775B1 (en) 2015-01-28 2015-10-13 Teva Pharmaceutical Industries, Ltd. Process for manufacturing glatiramer acetate product
WO2016128957A1 (en) * 2015-02-12 2016-08-18 Insight Biopharmaceuticals Ltd. Methods of determining relative potency of glatiramer acetate and use thereof in preparing a batch of glatiramer acetate as acceptable for pharmaceutical use
US11167003B2 (en) 2017-03-26 2021-11-09 Mapi Pharma Ltd. Methods for suppressing or alleviating primary or secondary progressive multiple sclerosis (PPMS or SPMS) using sustained release glatiramer depot systems

Also Published As

Publication number Publication date
US20120309671A1 (en) 2012-12-06
WO2003048735A3 (en) 2003-09-18
IL198285A (en) 2013-07-31
US20150094488A1 (en) 2015-04-02
US20110189706A1 (en) 2011-08-04
IS7286A (is) 2004-05-26
MXPA04005433A (es) 2004-10-11
US7429374B2 (en) 2008-09-30
ES2349033T3 (es) 2010-12-22
IL162127A (en) 2010-11-30
ZA200404472B (en) 2005-11-30
CN1618018A (zh) 2005-05-18
EP1459065B1 (en) 2010-07-28
CA2469393C (en) 2010-05-25
US20090053253A1 (en) 2009-02-26
US7923215B2 (en) 2011-04-12
NZ533327A (en) 2006-11-30
IL162127A0 (en) 2005-11-20
NO20042816L (no) 2004-09-06
EP1459065A4 (en) 2008-08-27
CY1110878T1 (el) 2015-06-10
JP2005511060A (ja) 2005-04-28
AU2002353059A1 (en) 2003-06-17
KR100657048B1 (ko) 2006-12-12
ATE475883T1 (de) 2010-08-15
US20030170729A1 (en) 2003-09-11
US20160025707A1 (en) 2016-01-28
EP1459065A2 (en) 2004-09-22
NO338247B1 (no) 2016-08-08
DE60237170D1 (de) 2010-09-09
CN1308683C (zh) 2007-04-04
HK1070421A1 (en) 2005-06-17
IL198285A0 (en) 2011-07-31
DK1459065T3 (da) 2010-10-11
SI1459065T1 (sl) 2010-11-30
US8389228B2 (en) 2013-03-05
AU2002353059B2 (en) 2008-06-19
PT1459065E (pt) 2010-10-11
CA2469393A1 (en) 2003-06-12
KR20050044676A (ko) 2005-05-12
JP4369234B2 (ja) 2009-11-18

Similar Documents

Publication Publication Date Title
US8389228B2 (en) Process for the measurement of the potency of glatiramer acetate
Shigehara et al. IL-12 and IL-18 are increased and stimulate IFN-γ production in sarcoid lungs
Cunningham-Rundles et al. Deficient IL-12 and dendritic cell function in common variable immune deficiency
Choi et al. Macrophage inflammatory protein 1-alpha is a potential osteoclast stimulatory factor in multiple myeloma
Rapoport et al. Decreased secretion of Th2 cytokines precedes up-regulated and delayed secretion of Th1 cytokines in activated peripheral blood mononuclear cells from patients with insulin-dependent diabetes mellitus
Jones et al. Reduced in vitro production of interferon-gamma, interleukin-4 and interleukin-12 and increased production of interleukin-6, interleukin-10 and tumour necrosis factor-alpha in systemic lupus erythematosus. Weak correlations of cytokine production with disease activity
Viallard et al. Analysis of leukemia inhibitory factor, type 1 and type 2 cytokine production in patients with eosinophilic fasciitis.
Agarwal et al. Pregnancy ameliorates induction and expression of experimental autoimmune uveitis
Linder et al. Pityrosporum orbiculare‐reactive T‐cell lines in atopic dermatitis patients and healthy individuals
Tsiavou et al. Intracellular IFN-γ production and IL-12 serum levels in latent autoimmune diabetes of adults (LADA) and in type 2 diabetes
Kakiuchi-Kiyota et al. Preclinical pharmacology and safety of RO7297089, a novel anti-BCMA/CD16a bispecific antibody for the treatment of multiple myeloma
Dabrowski et al. Competition of IL-1 and IL-1ra determines lymphocyte response to delayed stimulation with PHA.
Wolday et al. HIV‐1 Inhibits Leishmania‐lnduced Cell Proliferation but not Production of Interleukin‐6 and Tumour Necrosis Factor Alpha
Zhao et al. S-antigen specific T helper type 1 response is present in Behcet’s disease
Moazezi et al. Evaluation of immunological parameters in diabetic patients: are these patients immunodeficient?
Zhang et al. Circulating CXCR3-CCR6-CXCR5+ CD4+ T cells are associated with acute allograft rejection in liver transplantation
US20080138833A1 (en) Methods and compositions for assessing il-12 or the neutralization of il-12 in a sample
González-Rodríguez et al. Involvement of CD4+ and CD8+ T-lymphocytes in the modulation of nociceptive processing evoked by CCL4 in mice
US20050152914A1 (en) Formulation of antigen
Nomura et al. Spontaneous and Lipopolysaccharide-Stimulated Secretion of Cytokines by Peripheral Blood Monocytes in IgA Nephropathy Is Inhibited by lnterleukin-10
Li et al. Immunogenicity and toxicokinetics assessment of the mono-PEGylated recombinant human interleukin-11 in cynomolgus monkeys
Abdullah et al. Study the Correlation between Serum level of Interlukin 6 and Lactate Dehydrogenase in Myeloma Patients at Stage III with Serum Creatinine and Calcium Level
Gerenova et al. Hashimoto’s disease-involvement of cytokine network and role of oxidative stress in the severity of hashimoto’s thyroiditis
RU2555344C9 (ru) Способ лечения заболеваний, ассоциированных со сниженной продукцией bdnf
Crawley et al. Soluble IL-7R

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 162127

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2003549881

Country of ref document: JP

Ref document number: 533327

Country of ref document: NZ

Ref document number: 1020047008518

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2469393

Country of ref document: CA

Ref document number: PA/a/2004/005433

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2004/04472

Country of ref document: ZA

Ref document number: 200404472

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 1752/DELNP/2004

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2002353059

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2002790028

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 20028277732

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2002790028

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 198285

Country of ref document: IL